You are on page 1of 15

NIH Public Access

Author Manuscript
Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Published in final edited form as:
NIH-PA Author Manuscript

Int J Obes (Lond). 2010 October ; 34(0 1): S47–S55. doi:10.1038/ijo.2010.184.

Adaptive thermogenesis in humans


Michael Rosenbaum and Rudolph L. Leibel
Columbia University, College of Physicians & Surgeons, Departments of Pediatrics and Medicine,
Division of Molecular Genetics, New York, NY, 10032

Abstract
The increasing prevalence of obesity and its co-morbidities reflects the interaction of genes that
favor the storage of excess calories as fat with an environment that provides ad libitum availability
of calorically dense foods and encourages an increasingly sedentary lifestyle. While weight
reduction is difficult in and of itself, anyone who has every lost weight will confirm that it is much
harder to keep the weight off once it has been lost. The over 80% recidivism rate to pre-weight
loss levels of body fatness after otherwise successful weight loss is due to the coordinate actions
NIH-PA Author Manuscript

of metabolic, behavioral, neuroendocrine, and autonomic responses designed to maintain body


energy stores (fat) at a CNS- defined “ideal”. This “adaptive thermogenesis” creates the ideal
situation for weight regain and is operant in both lean and obese individuals attempting to sustain
reduced body weights. Much of this opposition to sustained weight loss is mediated by the
adipocyte-derived hormone “leptin”. The multiplicity of systems regulating energy stores and
opposing the maintenance of a reduced body weight illustrate that body energy stores in general
and obesity in particular are actively “defended” by interlocking bioenergetic and neurobiological
physiologies. Important inferences can be drawn for therapeutic strategies by recognizing obesity
as a disease in which the human body actively opposes the “cure” over long periods of time
beyond the initial resolution of symptomatology.

Keywords
Obesity; Adaptive Thermogenesis; Thyroid; Autonomic; Weight Loss

Introduction
Western societies tend to regard obesity, and the inability of individuals to sustain weight
NIH-PA Author Manuscript

loss, as largely self-imposed conditions which reflect a lack of “will power” related to
lifestyle changes. Optimal levels of adiposity are often defined by cosmetic rather than
medical considerations. However, genetic, epidemiological, and physiological studies
indicate that body fatness/weight is regulated, and that the increasing prevalence of obesity
in western societies reflects the interactions of genes favoring energy conservation and
storage with an environment which enables access to food calories and a more sedentary
lifestyle.

Throughout most of human evolution the tendency to store calories as fat would likely have
conferred an advantage by enabling survival during periods of prolonged caloric restriction,
as well as providing greater energy stores to nourish mother and fetus during and following
pregnancy. Thus, it is likely that the human genome would be enriched for alleles of genes
favoring the storage of calories as adipose tissue 1. Our current environment enables the

Address correspondence to: Michael Rosenbaum, M.D. Division of Molecular Genetics, Russ Berrie Medical Science Pavilion, 6th
Floor, 1150 St. Nicholas Avenue, New York, NY 10032, Tel: 212-305-9949, Fax: 212-304-5210, mr475@columbia.edu.
Rosenbaum and Leibel Page 2

consumption of large quantities of calorically dense foods and the maintenance of an


increasingly sedentary lifestyle. Clearly not everyone has the same “genetic risk” for obesity
and, regardless of any genetic proclivities, anyone will gain weight if they consume more
NIH-PA Author Manuscript

calories than they expend. As illustrated by diet-induced obesity (DIO)-prone and DIO-
resistant mouse strains 2, as well as humans3, 4, there is a heritable variability in the degree
of weight gain that different individuals will experience in an adipogenic environment. This
variability reflects, in part, heritable influences on how much an individual participates in
such an environment by increasing energy intake and/or decreasing energy expenditure, and
their metabolic responsiveness to an increase in energy intake relative to expenditure 5.

Any change in the amount of energy stored, predominantly as adipose tissue (over 100,000
kcal in a 70-kg man) but also as protein and glycogen, must reflect a difference between
energy taken in as food and energy expended in various forms of metabolic and physical
work (see below). If energy intake and output were not regulated by interlocking control
mechanisms that work concordantly to maintain energy stores, then a very small persistent
change in input relative to output would, over time, lead to substantial gain or loss of stored
calories. Yet, the average U.S. adult gains only 500–1000 g of weight (approximately 2000–
2500 kcal of stored energy) per year (more pronounced in older individuals, African-
Americans, Native-Americans, and Hispanic-Americans) 6, despite ingestion of
approximately 900,000–1,000,000 kcal/year. The remarkable constancy of body weight in
this context, presumably without conscious constant calculation of how many calories are
NIH-PA Author Manuscript

being consumed and/or expended by most individuals, suggests that energy intake and
expenditure vary directly to maintain relatively stable energy stores 7.

Metabolic responses to attempts to sustain weight loss


In long-term studies of weight-reduced children and adults, 80%-90% return to their
previous weight percentiles 8, while studies of those successful at sustained weight loss
indicate that the maintenance of a reduced degree of body fatness will probably require a
lifetime of meticulous attention to energy intake and expenditure 9, 10. The inability of most
otherwise successfully weight-reduced individuals to sustain weight loss reflects the actions
of potent and redundant metabolic, neuroendocrine, and autonomic systems (see below).

The responses of lean and obese individuals to experimental perturbations of body weight
suggest that the magnitude of stored energy, particularly fat, is defended by central nervous
system-mediated mechanisms that are similar, if not identical in lean and obese individuals.
In both lean and obese individuals, there is potent “opposition” to the maintenance of
reduced body weight that is achieved by coordinated regulation of energy intake and
expenditure mediated by signals emanating from adipose, gastrointestinal, and endocrine
tissues, and integrated by the liver and by central nervous system (see Table 1).
NIH-PA Author Manuscript

Energy expenditure
Maintenance of a 10% or greater reduction in body weight in lean or obese individuals is
accompanied by an approximate 20%-25% decline in 24-hour energy expenditure. This
decrease in weight maintenance calories is 10–15% below what is predicted solely on the
basis of alterations in fat and lean mass 11, 12. Thus, a formerly obese individual will require
~300–400 fewer calories per day to maintain the same body weight and physical activity
level as a never-obese individual of the same body weight and composition. Studies of
individuals successful at sustaining weight loss indicate that reduced weight maintenance
requires long-term lifestyle alterations 9. The necessity for these long-term changes is
consistent with the observation that the reduction in twenty four hour energy expenditure
(TEE) persists in subjects who have sustained weight loss for extended periods of time (6

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 3

months – 7 years) in circumstances of enforced caloric restriction in the biosphere 2


project 13, bariatric surgery 14 and lifestyle modification 15.
NIH-PA Author Manuscript

Twenty-four hour energy expenditure (TEE) is the sum of resting energy expenditure (REE;
cardiorespiratory work and the work of maintaining transmembrane ion gradients at rest;
approximately 60% of TEE), the thermic effect of feeding (TEF; the work of digestion;
approximately 5–10% of TEE), and non-resting energy expenditure (NREE, energy
expended in physical activity above resting; approximately 30–40% of TEE). The effects of
maintenance of reduced weight on each of these compartments of energy expenditure are
distinctly different. There is no significant decline in TEF following weight loss 11. Some
studies 16–18 report no change in REE following weight loss, while in others the
maintenance of a reduced body weight is associated with modest reductions in REE
accounting for about 10–15% of the decline in TEE beyond that predicted on the basis of
body composition changes 11, 12, 19. The variability in study results probably reflects
differences among studies in multiple factors including degree and duration of weight
stability before and after weight loss as well as changes in subject fitness and time spent in
physical activity following weight loss. Regardless of whether or not changes in REE
account for 10–15% of the changes in TEE following weight loss, NREE is clearly the
compartment of energy expenditure that is most affected by changes in body weight 11, 20
consistent with the importance of physical exercise in the successful maintenance of reduced
weight 9, 21.
NIH-PA Author Manuscript

The pre-eminence of NREE - accounting for as much of 85–90% of the decline in TEE
below predicted values in weight-reduced subjects 20, 22 could be due to declines in the
actual amount of physical activity performed. In rodents, maintenance of a reduced body
weight is associated with an increase, rather than decrease, in the amount of time spent in
physical activity 23, probably reflecting food-seeking behavior. In-patient and out-patient
studies of humans following weight loss have reported, respectively, no change or as much
as a 30% increase in the amount of time that subjects spend moving each day, 11, 18
supporting the view that skeletal muscle work efficiency is increased 20 (as opposed to
decreased amount of motion per se) following weight loss. Since these effects are most
evident at low levels of work, i.e., those commensurate with activities of daily living, it is
reasonable to infer that some of the opposition to reduced weight maintenance can be
diminished by exercising at higher levels of power output 20, 24.

Studies of skeletal muscle chemomechanical efficiency (calories expended above resting per
unit of power generated) in weight-reduced subjects indicate that maintenance of a reduced
body weight is associated with an approximate 20% increase in skeletal muscle work
efficiency at low levels of exercise, whether measured by bicycle ergometry or 31P-NMR
NIH-PA Author Manuscript

muscle spectroscopy 20. Ergometric studies measure whole body energy expenditure during
stationary cycling. Energy efficiency is expressed as kcal consumed above REE per unit of
power generated. Fuel utilization (fatty acid vs. glucose oxidation) is assessed by the
respiratory exchange ratio (RER, ratio of CO2 produced to O2 consumed). In 31P-NMR
spectroscopy, the ratio of inorganic phosphate (Pi), which increases during exercise due to
the hydrolysis of ATP, to phosphocreatine (PCr) which decreases during exercise to
replenish ATP, during exercise, reflects the efficiency of muscle in generating a specific
amount of power. In addition, the resting Pi reflects the relative fatty acid to glucose
oxidative potential of muscle, and the phosphocreatine recovery constant (kPCr, a constant
reflecting the exponential rate of PCr resynthesis following exercise as well the maximal
rate of oxygen consumption by muscle) reflects the muscle glycolytic potential 20, 25. 31P-
NMR spectroscopy can also be used to examine the in vivo ATP cost of single muscle
contraction by measuring the PCr depletion rate following muscle stimulation in an ischemic
limb (no PCr repletion until blood flow is restored) 26. Both of these methods demonstrate

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 4

that the maintenance of a 10% reduced body weight is associated with an approximate 20%
increase in skeletal muscle chemomechanical efficiency and an approximate 18% relative
increase in the fractional use of free fatty acids as fuel during low level exercise 20, 27 (see
NIH-PA Author Manuscript

Table 2). These results are consistent with studies of vastus lateralis muscle biopsies in
which the ratio of glycolytic (phosphofructokinase, PFK) to oxidative (cytochrome oxidase)
enzyme activities is significantly decreased following weight loss. The changes in these
enzyme ratios are sufficient to account in statistical analyses for a significant fraction of the
increased efficiency (R2=0.57, p<0.001) and free fatty acid oxidation (R2=0.31, p<0.01) that
occurs during low level-exercise in weight-reduced subjects 27.

Neuroendocrine Function
The neuroendocrine changes associated with the maintenance of a reduced body weight
include increased activity of the hypothalamic-pituitary-adrenal (HPA) axis and decreased
activity of the hypothalamic-pituitary-thyroid (HPT) and hypothalamic-pituitary-gonadal
(HPG) axes. The hypothalamic pro-opiomelanocortin (POMC)-melanocortin-melanocortin 4
receptor (MC4R) pathway, by virtue of its constituent neuronal outflow tracts to the ANS,
neuroendocrine axes, and cortical tracts subserving food intake, may provide a central nexus
for the sum of the integrated effects on energy expenditure and intake that are seen
following weight loss 1, 28, 29.

The importance of the HPA axis in regulating body fat stores is illustrated by the effects of
NIH-PA Author Manuscript

adrenalectomy on genetically obese rodents. The leptin-deficient or leptin-resistant mouse is


hyperphagic, hypometabolic (much like the weight-reduced human as discussed below),
hypercortisolemic (unlike leptin-deficient and resistant humans), and severely obese. These
phenotypes are abolished by chemical or surgical adrenalectomy 30. Hypercortisolemia
results in loss of lean body mass and increases the partitioning of stored calories to fat 1.

Studies of the HPA axis in which human subjects were assessed following variable weight
loss regimens and lengths of time maintaining a reduced weight have found increases 31,
decreases 32, and no change 33 in indices of cortisol production following weight loss.
Discrepancies among studies may reflect differences in subject populations regarding
exercise, gender, age, or weight loss regimens, as well as the degree of weight stability at the
time of study.

Thyroid hormone increases energy expenditure by increasing heart rate, blood pressure,
muscle ATP consumption (largely by stimulating production of muscle ATPase). The
thyroid hormone deficient patient is hypotensive, bradycardic, and lethargic and tends to
gain weight while the hyperthyroid patient is hypertensive and tachycardic and tends to lose
weight 34, 35. Both weight loss and the maintenance of a reduced body weight are associated
NIH-PA Author Manuscript

with small but statistically significant decreases in circulating concentrations of


triiodothyronine (T3) and increases in the circulating concentrations of its bioinactive
enantiomer reverse T3 (rT3), suggesting that weight loss results in increased peripheral
conversion of thyroxine (T4) to rT3 36. Thyroid releasing hormone (TRH)-stimulated
pituitary thyroid stimulating hormone (TSH) release is not diminished during caloric
restriction 37 or after weight loss 38 in humans. The lack of increase in TSH with weight
loss, despite the decrease in circulating concentrations of T3, indicates that hypothalamic
TRH release is decreased following weight loss. Caloric restriction and maintenance of a
reduced body weight are associated with decreased circulating leptin concentrations (see
below) 39. Low ambient leptin, in turn, reduces POMC production in hypothalamic neurons.
Decreased hypothalmic alpha-MSH (α-MSH), a proteolytic product of POMC, results in
decreased activity of hypothalamic pro-thyroid releasing hormone (pro-TRH) neurons in
rats 40, thus providing a possible mechanism for the decrease in HPT axis activity following

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 5

weight loss and the restoration of circulating concentrations of bioactive thyroid hormones
following leptin replacement 41.
NIH-PA Author Manuscript

Autonomic Nervous System Function


The autonomic nervous system (both parasympathetic and sympathetic) includes major
outflow tracts linking afferent biochemical signals regarding energy stores and efferent
tracts regulating energy expenditure. Increased parasympathetic nervous system activity
slows heart rate and decreases resting energy expenditure. The sympathetic branch of the
autonomic nervous system modulates feeding behavior, directly increases heart rate, and
acts directly on the thyroid gland to increase the rate of secretion of thyroid hormone 42, 43.
Sympathetic denervation of the arm in humans with palmar hyperhidrosis improves skeletal
muscle work efficiency 44 in arm muscles, and chemical sympathectomy attenuates leptin-
mediated increases in energy expenditure in rats 45. Daily urinary norepinephrine excretion
accounts for a significant proportion of the variance in energy expenditure and its
subcomponents in weight stable subjects 36.

The maintenance of a reduced body weight is associated with significant declines in SNS
tone (by analysis of heart rate following sequential parasympathetic and sympathetic
blockade or 24-hour urinary catecholamine excretion) and increases in PNS tone 36, 43, 46.
Changes in ANS tone associated with weight loss, in particular the decline in sympathetic
nervous system tone, may account for a significant fraction of the hypometabolic state
NIH-PA Author Manuscript

through direct effects on skeletal muscle, and/or indirectly via effects on circulating
concentrations of thyroid hormones36, 47, 48. Thus, weight-loss mediated changes in
autonomic nervous system activity may constitute a link between weight-loss associated
changes in energy and neuroendocrine homeostasis.

Brown Adipose Tissue


Brown adipose tissue (BAT) allows the uncoupling of mitochondrial substrate oxidation
from ATP production thereby releasing the energy of fatty acid oxidation as heat 49. This is
achieved via a 32kd “uncoupling protein” (UCP1) which is present in BAT, but not in white
adipose tissue (WAT). BAT has a rich sympathetic nerve and vascular supply and, in the
presence of cold, weight gain, and/or sympathetic nervous stimulation in rodents or
exogenous or endogenous hypercatecholaminemia in humans, BAT activity increases
resulting in heat generation 50, 51 The activation of BAT is dependent upon integration of
input from the sympathetic nervous system activation of adrenoreceptors (predominantly
β3) 52, with activation of at least one of the thyroid hormone receptors (TR) subtypes (TRα
or TRβ) 53. The leptin-sensitive declines in sympathetic nervous system activity and
circulating concentrations of bioactive thyroid hormones following weight loss that are
NIH-PA Author Manuscript

described above constitute a mechanism by which reduced obligatory and/or facultative


thermogenesis by BAT could contribute to adaptive thermogenesis in humans. As little as
25gm of BAT going from a maximally active to a minimally active state following weight
loss would be more than sufficient to account for the magnitude of decline in REE in
weight-reduced subjects that occurs beyond that predicted solely on the basis of weight and
body composition changes 54. Therefore, it is possible that a significant fraction of the
unexplained variance in resting energy expenditure or in changes in resting energy
expenditure following weight loss is attributable to changes in the activity or brown adipose
tissue (BAT) 55.

However, while BAT is a major contributor to adaptive thermogenesis in small mammals 56,
its role in thermogenesis in adult humans remains unclear. In rodents, BAT contributes to
both obligatory thermogenesis (the heat produced to maintain body temperature at rest) and
facultative thermogenesis (the heat produced to maintain body temperature at ambient

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 6

temperatures below thermoneutrality) 53. The cold intolerance of hypothyroid rodents


reflects declines in both obligatory and facultative thermogenesis by BAT 53. BAT is easily
detected in rodents and clearly plays a role in non-shivering thermogenesis in human
NIH-PA Author Manuscript

neonates.

Previous studies showed a lack of a significant presence of BAT in humans except under
extreme conditions of hypercatecholaminemia 57 and, until recently, no studies have been
carried out quantifying the contribution of BAT to total adaptive thermogenesis in humans.
Recent advances in positive emission tomography (PET) scanning technology have allowed
detailed imaging of BAT using uptake of 2-[18F]fluoro-2-desoxy-glucose (FDG) and a
hybrid scanner. FDG uptake has been shown to correspond to the neck, supraclavicular,
mediastinal, paraspinal, paravertebral and renal areas known to contain BAT in
humans 58,59, 60 and FDG uptake in these areas (www.med.harvard.edu/JPNM/chetan/
normals/brown_fat/case.html) is inhibited by β-adrenergic blockade with propranolol 61–63.
In a recent series of papers 54, 64, 65, several groups demonstrated the ability to detect BAT
in healthy human beings with varying results as to whether thermal stimuli are necessary to
detect it. In a retrospective study of [18F]FDG (dose not specified) PET scans, Cypress et
al 54 detected BAT in 7.5% of women and 3.1% of men studied under thermoneutral
conditions. Lichtenbelt et al 64 found that BAT was detected in 23/24 subjects after cold
exposure (16°C for 2 hours) but was not detected in 3 of these subjects who were also
studied under thermoneutral conditions([18F]FDG dose of 74 MBq). Finally, Virtanen et
NIH-PA Author Manuscript

al 65 reported that BAT was detected in 5/5 adult subjects under both thermoneutral and
post-cold exposure (19°C for 2 hours) conditions using a higher dose (185 MBq) of
[18F]FDG.

The anatomic identification of BAT in humans using FDG does not necessarily reflect actual
thermogenic activity of BAT, and the question remains as to whether BAT actually
participates in resting thermogenesis, diet-induced thermogenesis, or adaptive thermogenesis
following weight loss or gain in humans. Increased glucose uptake by BAT is considered to
reflect increased metabolic activity and thermogenesis 56, 66, 67. This is the basis for the use
of the FDG PET technique in localization of tumors and for the analysis of brain areas
involved in different cognitive activities as well as for examining myocardial metabolic
activity56. Thus, the FDG uptake seen in brown adipose tissue in adult man implies the
existence of thermogenically active tissue in adult man. The magnitude of glucose uptake by
BAT in FDG PET studies of subjects fasted and at rest, compared to other tissues, also
suggests that BAT may play a significant role in glucose disposal in low activity states (i.e.,
the state in which we spend at least 1/3 of our lives). Assuming that reduced BAT activation
following weight loss is a significant factor in adaptive thermogenesis, this effect is more
likely to be evident in obligatory than facultative thermogenesis. In this environment we
NIH-PA Author Manuscript

spend almost all of our time in thermoneutral conditions reducing the need for facultative
thermogenesis and possibly contributing to the increasing prevalence of obesity 68, 69.
Further studies of the role of BAT in human thermogenesis outside of the neonatal period
are clearly indicated.

Metabolic responses to attempts to sustain weight gain


The metabolic changes that occur in subjects during maintenance of an elevated body weight
following overfeeding involve many of the same systems but are not, in fact, mirror images
of the changes following weight loss. In a manner complementary to that seen following
weight loss, the maintenance of an elevated body weight is associated with significant
increases in circulating concentrations of T3 and T4, SNS tone, TEE, NREE, and, of course,
circulating leptin concentrations and a decreases in PNS tone and skeletal muscle work
efficiency. However, there is no demonstrable effect of the maintenance of an elevated body
weight on circulating concentrations of TSH, and there is a much more marked effect of

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 7

elevated weight maintenance on TEF and less of an effect on REE than is seen following
weight loss11, 36, 70.
NIH-PA Author Manuscript

Unlike the metabolic opposition to sustaining a reduced body weight, which persists long
after weight reduction in mice 71 and humans 72, the increased energy expenditure noted
during short-term overfeeding in mice seems to be short-lived. Rodents with diet-induced
obesity demonstrate increased energy expenditure 73 and increased SNS tone74 during the
first 3–4 weeks of overfeeding. However, after a few months on a high fat diet, these
changes are no longer evident 74, 75, indicating that resistance to sustained increased
adiposity is less sustained than resistance to decreased adiposity 69. The steadily increasing
prevalence of obesity in humans also suggests that body fatness is facilitated more
vigorously than body thinness. In addition to the lack of physiological persistence of strong
metabolic opposition to weight gain, any “defense” against further weight gain is stretched
to the limit by this lifestyle, while opposition to sustaining weight loss remains potent and
viable76.

Energy Intake
As noted above, the long-term constancy of body weight suggests that energy intake and
expenditure vary coordinately to maintain relatively stable energy stores. This “coupling”
which reduces caloric intake in response to decreased energy expenditure is disrupted during
and following weight loss 7. During dynamic weight loss, human beings and rodents are
NIH-PA Author Manuscript

both hungrier (willing to eat more often) and less satiated (willing to eat more per meal) 77.
Even during maintenance of a reduced weight, satiety remains diminished despite the
decline in energy expenditure 78. The simultaneous declines in both energy expenditure and
satiety following weight loss conspire to create the optimal biological circumstance for
weight regain.

Leptin in Energy Homeostasis


A critical mediator of these reciprocal changes in energy intake and expenditure is the
hormone leptin. Leptin is an adipocyte derived molecule that circulates in weight-stable
individuals in direct proportion to fat mass 79. The hyperphagic, hypometabolic phenotype
of weight-reduced humans is similar to that of leptin-deficient or -unresponsive humans and
rodents 80. Circulating leptin concentrations are inversely correlated with hunger ratings in
humans during weight loss, independent of the amount of weight or body fat lost 81. Leptin
administration reverses the hyperphagia associated with leptin deficiency in leptin-deficient
mice and humans 82, 83, and acts synergistically with sibutramine to reduce food intake in
rodents 84. Leptin suppresses food intake by promoting the production of anorexigenic
neuropeptides (processed products of POMC) and reducing the expression of orexigens such
NIH-PA Author Manuscript

as neuropeptide Y (NPY), agouti-related peptide (AgRP), and melanin concentrating


hormone (MCH). Thus, decreased circulating leptin concentrations as a result of reduced fat
mass has the net effect of stimulating food intake 1.

The hypothalamic POMC-melanocortin-MC4R pathway is highly sensitive to circulating


leptin concentrations and POMC expression is decreased in low-leptin states 29, 85. Briefly,
POMC is cleaved to alpha-melanocyte stimulating hormone (α-MSH) and beta-endorphin
(β-EP) as well as other bioactive molecules. As discussed previously, α-MSH stimulates
release of hypothalamic pro-TRH. β-EP inhibits the release of hypothalamic corticotropin
releasing factor (CRF). Therefore, reduced ambient leptin induced by weight loss, should be
associated with decreased HPT and increased HPA axis activity. Mice overexpressing the
melanocortin 4 receptor (MC4R) antagonists agouti signaling protein (ASP) or agouti-
related peptide (AgRP) 86- as well as rodents and humans with hypomorphic mutations in
MC4R 87, disruptions of POMC gene expression 88, 89 or of proproneuropeptide (e.g.,

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 8

POMC, pro-ACTH, pro-TRH) processing by prohormone convertases 90, 91 - are obese. The
importance of leptin in mediating these effects is reflected in the observation that fasting in
rodents causes hypoleptinemia that is associated with increased arcuate and brainstem NPY
NIH-PA Author Manuscript

and AgRP mRNA expression and decreased POMC mRNA in lean animals, but not in
leptin-receptor deficient animals 92.

Administration of leptin to leptin-deficient rodents and humans in doses that restore


circulating leptin concentrations to their physiological range increases energy
expenditure 93, decreases energy intake, increases sympathetic nervous system activity 94,
and normalizes hypothalamic-pituitary-adrenal, thyroid, and gonadal function1, 29, 82. Yet, in
humans (lean or obese) and rodents who are not leptin-deficient, induction of weight loss
requires doses of leptin that produce plasma leptin concentrations over 10 times
normal 95, 96. Recent studies of the short-term administration of leptin to weight-reduced
lean and obese subjects suggest that restoration of circulating concentrations of leptin to
levels present prior to weight loss reverses the decreased energy expenditure, and its
associated declines in thyroid hormone and SNS activity and increase in skeletal muscle
work efficiency, and increased energy intake, measured behaviorally and by functional
magnetic resonance imaging of neuronal responses to food, that characterize the weight-
reduced state78, 82, 97. In this sense, the weight-reduced state may be perceived by CNS
components relevant to energy homeostasis as a state of relative leptin deficiency.
Pharmacotherapy activating the leptin-signaling pathway may help weight-reduced
NIH-PA Author Manuscript

individuals to sustain their weight loss98.

Summary
Attempts to sustain weight loss invoke adaptive responses involving the coordinate actions
of metabolic, neuroendocrine, autonomic, and behavioral changes that “oppose” the
maintenance of a reduced bodyweight. This phenotype is distinct from that opposing
dynamic weight loss per se. The multiplicity of systems regulating energy stores and
opposing the maintenance of a reduced body weight illustrate that body energy stores in
general and fat stores in particular are actively “defended” by interlocking bioenergetic and
neurobiological physiologies. Important inferences can be drawn for therapeutic strategies
by recognizing obesity as a state in which the human body actively opposes the “cure” over
long periods of time beyond the initial resolution of symptomatology.

References
1. Leibel, R.; Chua, S.; Rosenbaum, M. Chapter 157. Obesity. In: Scriver, C.; Beaudet, A.; Sly, W.;
Valle, D., editors. The metabolic and molecular bases of inherited disease. 8. Vol. III. New York:
NIH-PA Author Manuscript

McGraw-Hill; 2001. p. 3965-4028.


2. West DB, Boozer CN, Moody DL, Atkinson RL. Dietary obesity in nine inbred mouse strains. Am J
Physiol. 1992; 262:R1025–R32. [PubMed: 1621856]
3. Bouchard C, Tremblay A. Genetic influences on the response of body fat and fat distribution to
positive and negative energy balances in human identical twins. J Nutr. 1997; 127:943S–47S.
[PubMed: 9164270]
4. Bouchard C, Tremblay A, Despres JP, Nadeau A, Lupien PJ, Theriault G, et al. The response to
long-term overfeeding in identical twins. N Engl J Med. 1990; 322:1477–82. [PubMed: 2336074]
5. Rosenbaum M. Epidemiology of pediatric obesity. Ped Annals. 2007; 36:89–95.
6. Lewis C, Jacobs D, McCreath H, Kiefe C, Schreiner P, Smith D, et al. Weight gain continues in the
1990s: 10-year trends in weight and overweight from the CARDIA study. Coronary Artery Risk
development in Young Adults. Am J Epidemiol. 2000; 151:1172–81. [PubMed: 10905529]
7. Moore M. Interactions between physical activity and diet in the regulation of body weight. Proc
Nutr Soc. 2000; 59:193–98. [PubMed: 10946787]

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 9

8. McGuire M, Wing R, Klem M, Hill J. Behavioral strategies of individuals who have maintained
long-term weight losses. Obes Res. 1999; 7:334–41. [PubMed: 10440589]
9. Wing R, Hill J. Successful weight loss maintenance. Annu Rev Nutr. 2001; 21:323–41. [PubMed:
NIH-PA Author Manuscript

11375440]
10. Klem M, Wing R, Lang W, McGuire M, Hill J. Does weight loss maintenance become easier over
time. Obes Res. 2000; 8:438–44. [PubMed: 11011910]
11. Leibel R, Rosenbaum M, Hirsch J. Changes in energy expenditure resulting from altered body
weight. N Eng J Med. 1995; 332:621–28.
12. Weigle D, Sande K, Iverius P, Monsen E, Brunzell J. Weight loss leads to a marked decrease in
nonresting energy expenditure in ambulatory human subjects. Metabolism. 1988; 37:930–36.
[PubMed: 3173112]
13. Weyer C, Walford R, Harper I, Milner M, MacCallum T, Tataranni P, et al. Energy metabolism
after 2 y of energy restriction: the biosphere 2 experiment. Am J Clin Nutr. 2000; 72:946–53.
[PubMed: 11010936]
14. Gemert, Wv; Westerterp, K.; Acker, Bv; Wagenmakers, A.; Halliday, D.; Greve, J., et al. Energy,
substrate and protein metabolism in morbid obesity before, during and after massive weight loss.
Int J Obes. 2000; 24:711–18.
15. Leibel R, Hirsch J. Diminished energy requirements in reduced-obese patients. Metabolism. 1984;
33:164–70. [PubMed: 6694559]
16. dePeuter R, Withers R, Brinkman M, Tomas F, Clark D. No differences in rates of energy
expenditure between post-obese women and their matched, lean controls. Int J Obes. 1992;
NIH-PA Author Manuscript

16:801–08.
17. Amatruda J, Statt M, Welle S. Total and resting energy expenditure in obese women reduced to
ideal body weight. J Clin Invest. 1993; 92:1236–42. [PubMed: 8376583]
18. Weinsier R, Hunter G, Zuckerman P, Redden D, Darnell B, Larson D, et al. Energy expenditure
and free-living physical activity in black and white women: comparison and after weight loss. Am
J Clin Nutr. 2000; 71:1138–46. [PubMed: 10799376]
19. Astrup A, Gotzsche P, Werken Kvd, Ranneries C, Toubro S, Raben A, et al. Meta-analysis of
resting metabolic rate in formerly obese subjects. Am J Clin Nutr. 1999; 69:1117–22. [PubMed:
10357728]
20. Rosenbaum M, Vandenborne K, Goldsmith R, Simoneau J, Heymsfield S, Joanisse D, et al. Effects
of experimental weight perturbation on skeletal muscle work efficiency in human subjects. Amer J
Physiol. 2003; 285:R183–92.
21. Klem M, Wing R, McGuire M, Seagle H, Hill J. A descriptive study of individuals successful at
long term maintenance of substantial weight loss. Am J Clin Nutr. 1998; 66:239–46. [PubMed:
9250100]
22. Newcomer B, Larson-Meyer D, Hunter G, Weinsier R. Skeletal muscle metabolism in overweight
and post-overweight women: an isometric exercise study using (31)P magnetic resonance
spectroscopy. Int J Obes. 2001; 25:1309–15.
NIH-PA Author Manuscript

23. Pirke K, Briicjs A, Wilckens T, Marquard R, Schweiger U. Starvation induced hyperactivity in the
rat: the role of endocrine and neurotransmitter changes. Neurosci Biobehav Rev. 1993; 17:287–94.
[PubMed: 7903806]
24. Freyschuss U, Melcher A. Exercise energy expenditure in extreme obewsity: influence of
ergometry type and weight loss. Scand J Clin Lab Invest. 1978; 38:753–59. [PubMed: 741204]
25. Walter G, Vandenborne K, McCully K, Leigh J. Noninvasive measurement of PCr recovery
kinetics in single human muscles. Am J Physiol. 1997; 268:C869–76. [PubMed: 7733235]
26. Blei M, Conley K, Kushmerick M. Separate measures of ATP utilization and recovery in human
skeletal muscle. J Physiol (Lond). 1993; 465:203–22. [PubMed: 8024651]
27. Goldsmith R, Joanisse D, Gallagher D, Pavlovich K, Shamoon E, Leibel R, et al. Effects of
experimental weight perturbation on skeletal muscle work efficiency, fuel utilization, and
biochemistry in human subjects. Am J Physiol. 2009 In Press.
28. Schwartz M, Woods S, Porte D, Seeley R, Baskin D. Central nervous system control of food
intake. Nature. 2000; 404:661–70. [PubMed: 10766253]

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 10

29. Wardlaw S. Clinical review 127: Obesity as a neuroendocrine disease: lessons to be learned from
proopiomelanocortin and melanocortin receptor mutations in mice and men. J Clin Endocrinol
Metab. 2001; 86:1442–6. [PubMed: 11297566]
NIH-PA Author Manuscript

30. Shimomura Y, Bray GA, Lee M. Adrenalectomy and steroid treatment in obese (ob/ob) and
diabetic (db/db) mice. Horm Metab Res. 1987; 19:295–99. [PubMed: 2957297]
31. Kennedy S, Brown G, McVrey G, Garfinkel P. Pineal and adrenal function before and after
refeeding in anorexia nervosa. Biol Psych. 1991; 30:216–24.
32. Guldstrand M, Ahren B, Wredling R, Backman L, Linus P, Adamson U. Alteration of the
counterregulatory responses to insulin-induced hypoglycemia and of cognitive function after
massive weight reduction in severely obese subjects. Metabolism. 2003; 52:900–07. [PubMed:
12870168]
33. Yanovski J, Yanovski S, Gold P, Chorousos G. Differences in corticotropin-releasing hormone-
stimulated adrenocorticotropin and cortisol before and after weight loss. J Clin Endocrinol Metab.
1997; 82:1874–78. [PubMed: 9177399]
34. Danforth E, Burger A. The role of thyroid hormones in the control of energy expenditure. Clin
Endocrinol Metab. 1984; 13:581–96. [PubMed: 6391756]
35. al-Adsani H, Hoffer L, Silva J. Resting energy expenditure is sensitive to small dose changes in
patients on chronic thyroid hormone replacement. J Clin Endocrinol Metab. 1997; 82:1118–25.
[PubMed: 9100583]
36. Rosenbaum M, Hirsch J, Murphy E, Leibel R. The effects of changes in body weight on
carbohydrate metabolism, catecholamine excretion, and thyroid function. Amer J Clin Nutr. 2000;
NIH-PA Author Manuscript

71:1421–32. [PubMed: 10837281]


37. Goodwin G, Fairburn C, Keenan J, Cowen P. The effects of dieting and weight loss upon the
stimulation of thyrotropin (TSH) by thyrotropin-releasing hormone (TRH) and suppression of
cortisol secretion by dexamethasone in men and women. J Affect Dis. 1988; 14:137–44. [PubMed:
2966827]
38. Guzzaloni G, Grugni G, Moro D, Calo G, Tonelli E, Ardizzi A, et al. Thyroid-stimulating hormone
and prolactin responses to thyrotropin-releasing hormone in juvenile obesity before and after
hypocaloric diet. J Encodrinol Invest. 1995; 18:621–29.
39. Rosenbaum M, Nicolson M, Hirsch J, Murphy E, Chu F, Leibel R. Effects of weight change on
plasma leptin concentrations and energy expenditure. J Clin Endocrinol Metab. 1997; 82:3647–54.
[PubMed: 9360521]
40. Fekete C, Legradi G, Mihaly E, Huang Q, Tatro J, Rand W, et al. alpha-Melanocyte-stimulating
hormone is contained in nerve terminals innervating thyrotropin-releasing hormone-synthesizing
neurons in the hypothalamic paraventricular nucleus and prevents fasting-induced suppression of
prothyrotropin-releasing hormone gene expression. J Neurosci. 2000; 20:1550–58. [PubMed:
10662844]
41. Harris M, Aschkenasi C, Elias C, Chandrankunnel A, Nillni E, Bjorbaek C, et al. Transcriptional
regulation of the thyrotropin-releasing hormone gene by leptin and melanocortin signaling. J Clin
Invest. 2001; 107:111–20. [PubMed: 11134186]
NIH-PA Author Manuscript

42. Lang C, Rayos G, Chomsky D, Wood A, Wilson J. Effect of sympathoinhibition on exercise


performance in patients with heart failure. Circulation. 1997; 96:238–45. [PubMed: 9236440]
43. Aronne L, Mackintosh R, Rosenbaum M, Leibel R, Hirsch J. Autonomic nervous system activity
in weight gain and weight loss. Am J Physiol. 1995; 38:R222–25.
44. Kardos A, Taylor D, Thompson C, Styles P, Hands L, Collin J, et al. Sympathetic denervation of
the upper limb improves forearm exercise performance and skeletal muscle bioenergetics.
Circulation. 2000; 13:2716–20. [PubMed: 10851209]
45. Dobbins R, Szczepaniak L, Zhang W, McGarry J. Chemical sympathectomy alters regulation of
body weight during prolonged ICV leptin infusion. Am J Physiol. 2003; 284:E778–87.
46. Aronne L, Mackintosh R, Rosenbaum M, Leibel R, Hirsch J. Cardiac autonomic nervous system
activity in obese and never-obese young men. Obes Res. 1997; 5:354–59. [PubMed: 9285844]
47. Kim M, Small C, Stanley S, Morgan D, Seal L, Kong W, et al. The central melanocortin system
affects the hypothalamo-pituitary thyroid axis and may mediate the effect of leptin. J Clin Invest.
2000; 105:1005–11. [PubMed: 10749579]

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 11

48. Flier J, Harris M, Hollenberg A. Leptin, nutrition, and the thyroid: the why, the wherefore, and the
wiring. J Clin Invest. 2000; 105:859–61. [PubMed: 10749565]
49. Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol
NIH-PA Author Manuscript

Rev. 2004; 84:277–359. [PubMed: 14715917]


50. Trayhurn, P.; Nicholls, D., editors. Brown Adipose Tissue. Baltimore, MD: Edward Arnold; 1986.
p. 117-23.
51. Leibel, R.; Berry, E.; Hirsch, J. In vivo evidence for catechol-responsive brown adipose tissue in
obese patients. Proceedings Fifth International Congress on Obesity; Jerusalem, Israel. 1987;
52. Chemogubova E, Hurchinson D, Nedergaard J, Bengtsson T. Alpha1- and beta1-adrenoceptor
signaling fully compensates for beta3-adrenoceptor deficiency in brown adipocyte norepinephrine-
stimulated glucose uptake. Endocrinology. 2005; 146:2271–84. [PubMed: 15665039]
53. Alkemade A. Central and peripheral effects of thyroid hormone signalling in the control of energy
metabolism. J Neuroendocrinol. 2010; 22:56–63. [PubMed: 19912477]
54. Cypress A, Lehman S, Williams G, Tal I, Rodman D, Goldfine A, et al. Identification and
importance of brown adipose tissue in adult humans. N Eng J Med. 2009; 360:1509–17.
55. Seale P, Lazar M. Brown fat in humans: turning up the heat on obesity. Diabetes. 2009; 68:1482–
84. [PubMed: 19564460]
56. Nedergaard, J.; Connolly, E.; Cannon, B. Brown adipose tissue in the mammalian neonate. In:
Trayhurn, P.; Nicholls, D., editors. Brown Adipose Tissue. Baltimore, MD: Edward Arnold; 1986.
p. 152-213.
57. Astrup A, Bulow J, Madsen J, Christensen N. Contribution of BAT and skeletal muscle to
NIH-PA Author Manuscript

thermogenesis induced by ephedrine in man. Am J Physiol. 1985; 248:E507–15. [PubMed:


3922230]
58. Nedergard J, Bengtsson T, Cannon B. Unexpected evidence for active brown adipose tissue in
adult humans. Am J Physiol. 2007; 293:E444–52.
59. Truong M, Erasmus J, Munden R, Marom E, Sabloff B, Gladish G, et al. Focal FDG uptake in
mediastinal brown fat mimicking malignancy: a potential pitfall resolved on PET/CT. Am J
Roentgenol. 2004; 183:1127–32. [PubMed: 15385319]
60. Chib C, Katsuragi I, Simada T, Adachi I, Satoh Y, Noguchi H, et al. Evaluation of human brown
adipose tissue using positron emission tomography, computerised tomography and histochemical
studies in association with body mass index, visceral fat accumulation and insulin resistance
(Abstract). Obes Rev. 2006; 7:87.
61. Jacobsson H, Bruzelius M, Larsson S. Reduction of FDG uptake in brown adipose tissue by
propranolol. Eur J Nucl Mol Imaging. 2005; 32:1130.
62. Tatsumi M, Engles J, Ishimori T, Nicely O, Cohade C, Wahl R. Intense (18)F-FDG uptake in
brown fat can be reduced pharmacologically. J Nucl Med. 2004; 45:1189–93. [PubMed:
15235065]
63. Soderlund V, Larsson S, Jacobsson H. Reduction of FGD uptake in brown adipose tissue in clinical
patients by a single dose of propranolol. Eur J Nucl Med Mol Imaging. 2007; 45:1018–22.
NIH-PA Author Manuscript

[PubMed: 17225118]
64. Lichtenbelt W, Vanjommeng J, Smulders N, Drossaerts J, Kemerink G, Bouvy N, et al. Cold-
activated brown adipose tissue in healthy men. N Eng J Med. 2009; 360:1500–08.
65. Virtanen K, Lidell M, Orava J, Jeglind M, Westergren R, Niemi T, et al. Functional brown adipose
tissue in healthy adults. N Eng J Med. 2009; 360:1518–25.
66. Chernogubova E, Cannon B, Bengtsson T. Norepinephrine increases glucose transport in brown
adipocytes via 3-adrenoceptors through a cAMP, PKA and PI3-kinase-dependent pathway
stimulating conventional and novel PKCs. Endocrinol. 2004; 145:269–80.
67. Marette A, Bukowiecki L. Noradrenaline stimulates glucose transport in rat brown adipocytes by
activating thermogenesis. Evidence that fatty acid activation of mitochondrial respiration enhances
glucose transport. Biochem J. 1991; 277:119–24. [PubMed: 1713031]
68. Elobeid M, Allison D. Putative environmental-endocrine disruptors and obesity: a review. Curr
Opin Endocrinol Diabetes Obes. 2008; 15:403–08. [PubMed: 18769210]
69. Schwartz M, Woods S, Seeley R, Barsh G, Baskin D, Leibel R. Is the energy homeostasis system
inherently biased toward weight gain? Diabetes. 2003; 52:232–38. [PubMed: 12540591]

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 12

70. Tremblay A, Despres J, Theriault G, Fournier G, Bouchard C. Overfeeding and energy expenditure
in humans. Am J Clin Nutr. 1992; 56:857–62. [PubMed: 1415004]
71. Dulloo A, Girardier L. 24 hour energy expenditure several months after weight loss in the underfed
NIH-PA Author Manuscript

rat: evidence for a chronic decrease in whole-body metabolic efficiency. Int J Obes. 1993; 17:115–
23.
72. Rosenbaum M, Hirsch J, Gallagher D, Leibel R. Long-term persistence of adaptive thermogenesis
in subjects who have maintained a reduced body weight. Amer J Clin Nutr. 2008; 88:906–12.
[PubMed: 18842775]
73. Rothwell N, Stock M. Energy expenditure of cafeteria-fed rats determined from measurements of
energy balance and indirect calorimetry. J Physiol London. 1982; 328:371–77. [PubMed:
7131317]
74. Levin BE, Triscari J, Sullivan AC. Altered sympathetic activity during development of diet-
induced obesity in rat. Am J Physiol. 1983; 244:R347–R55. [PubMed: 6131610]
75. Corbett SW, Stern JS, Keesey RE. Energy expenditure in rats with diet-induced obesity. Am J Clin
Nutr. 1986; 44:173–80. [PubMed: 3728354]
76. Murgatroyd P, Goldberg G, Leahy F, Prentice A. Effects of inactivity and diet composition on
human energy balance. Int J Obes. 1999; 23:1269–75.
77. Doucet E, Cameron J. Appetite control after weight loss: what is the role of bloodborne peptides?
Amer J Physiol Nutr Metab. 2007; 32:523–32.
78. Kissileff H, Thornton M, Torres M, Pavlovich K, Leibel R, Rosenbaum M. Maintenance of
reduced body weight in humans is associated with leptin-reversible declines in satiation. 2010
NIH-PA Author Manuscript

(submitted).
79. Rosenbaum M, Nicolson M, Hirsch J, Heymsfield S, Gallagher D, Chu F, et al. Effects of gender,
body composition, and menopause on plasma concentrations of leptin. J Clin Endocrinol Metab.
1996; 81:3424–27. [PubMed: 8784109]
80. Leibel R. The role of leptin in the control of body weight. Nutr Rev. 2002; 60:S15–19. [PubMed:
12403079]
81. Heini A, Lara-Castro C, Kirk K, Considine R, Caro J, Weinsier R. Association of leptin and
hunger-satiety ratings in obese women. Int J Obes. 1998; 22:1084–87.
82. Farooqi I, Jebb S, Langmack G, Lawrence E, Cheetham C, Prentice A, et al. Effects of
recombinant leptin therapy in a child with congenital leptin deficiency. N Eng J Med. 1999;
341:879–84.
83. Campfield LA, Smith FJ, Guisez Y, Devos R, Burn P. Recombinant mouse OB protein: evidence
for a peripheral signal linking adiposity and central neural networks. Science. 1995; 269:546–49.
[PubMed: 7624778]
84. Boozer C, Love R, Cha M, Leibel R, Aronne L. Synergy of leptin and sibutramine in treatment of
diet-induced obesity in rats. Metabolism. 2001; 50:889–93. [PubMed: 11474475]
85. Korner J, Chua S, WIlliams J, Leibel R, Wardlaw S. Regulation of hypothalamic pro-
opiomalanocortin by lean and obese rats. Neuroendocrinol. 1999; 70:377–83.
NIH-PA Author Manuscript

86. Ollmann M, Wilson B, Yang Y, Kerns J, Chen Y, Barsh G. Antagonism of central melanocortin
receptors in vitro and in vivo by agouti-related protein. Science. 1997; 278:135–38. [PubMed:
9311920]
87. Farooqi I, Keogh J, Yeo G, Lank E, Ceetham T, O’Rahilly S. Clinical spectrum of obesity and
mutations in the melanocortin 4 receptor gene. N Eng J Med. 2003; 348:1085–95.
88. Krude H, Biebermann H, Luck W, Horn R, Brabant G, Gruters A. Severe early-onset obesity,
adrenal insufficiency, and red hair pigmentation caused by POMC nutations in humans. Nat Genet.
1998; 19:155–57. [PubMed: 9620771]
89. Yaswen L, Diehl N, Brennan M, Hochgeswender U. Obesity in the mouse model of pro-
opiomelanocortin deficiency responds to peripheral melanocortin. Nat Med. 1999; 5:1066–70.
[PubMed: 10470087]
90. Naggert JK, Fricker LD, Varlamov O, Nishin RM, Rouille T, Steiner DF, et al.
Hyperproinsulinaemia in obese fat/fat mice associated with a carboxypeptidase E mutation which
reduces enzyme activity. Nature Genet. 1995; 10:135–42. [PubMed: 7663508]

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 13

91. Jackson RS, Creemers JWM, Ohagi S, Raffin-Sanson M-L, Sanders L, Montague CT, et al.
Obesity and impaired prohormone processing associated with mutations in the human prohormone
convertase 1 gene. Nature Genetics. 1997; 16:303. [PubMed: 9207799]
NIH-PA Author Manuscript

92. Korner J, Wardlaw S, Liu S, Conwell I, Leibel R, Chua S. Effects of leptin receptor mutation on
Agrp gene expression in fed and fasted lean and obese (LA/N-faf) rats. Endocrinol. 2000;
141:2465–71.
93. Solinas G, Summermatter S, Mainieri D, Gubler M, Pirola L, Wymann M, et al. The direct effect
of leptin on skeletal muscle thermogenesis is mediated by substrate cycling between de novo
lipogenesis and lipid oxidation. Febs. 2004; 577:539–44.
94. Satoh N, Ogawa Y, Katsuura G, Numata Y, Tsuji T, Hayase M, et al. Sympathetic activation of
leptin via the ventromedial hypothalamus: leptin-induced increase in catecholamine secretion.
Diabetes. 1999; 48:1787–93. [PubMed: 10480609]
95. Heymsfield SB, Greenberg AS, Fujioka K, Dixon RM, Kushner R, Hunt T, et al. Recombinant
leptin for weight loss in obese and lean adults: A randomized, controlled, dose-escalation trial.
JAMA. 1999; 292:1568–75. [PubMed: 10546697]
96. Campfield L, Smith F, Guisez Y, Devos R, Burn P. Recombinant mouse OB protein: Evidence for
a peripheral signal linking adiposity and central neural networks. Science. 1995; 269:546–48.
[PubMed: 7624778]
97. Rosenbaum M, Goldsmith R, Bloomfield D, Magnano A, Weimer L, Heymsfield S, et al. Low
dose leptin reverses skeletal muscle, autonomic, and neuroendocrine adaptations to maintenance of
reduced weight. J Clin Invest. 2005; 115:3579–86. [PubMed: 16322796]
NIH-PA Author Manuscript

98. Ravussin E, Smith S, Mitchell J, Shringarpure R, Shan K, Maier H, et al. Enhanced weight loss
with pramlintide/metreleptin: an integrated neurohormonal approach to obesity pharmacotherapy.
Obesity. 2009; 17:1736–43. [PubMed: 19521351]
99. Welt C, Chan J, Bullen J, Murphy R, Smith P, DePaoli A, et al. Recombinant human leptin in
women with hypothalamic amenorrhea. N Eng J Med. 2004; 351:987–97.
NIH-PA Author Manuscript

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 14

Table 1
Changes in energy expenditure, autonomic nervous system function, and neuroendocrine function in subjects
NIH-PA Author Manuscript

maintaining a reduced body weight with or without leptin “replacement” 1, 97, 99

Effects of 10% Reduced Weight Maintenance Effects of Leptin Administration to Weight-


Reduced Subjects

Energy Expenditure
Twenty-four-hour energy expenditure Decreased (−15%) Reversed

Resting energy expenditure Decreased or unchanged No significant change

Thermic effect of feeding Unchanged Unchanged

Non-resting energy expenditure Decreased (−30%) Reversed

Skeletal muscle work efficiency Increased (20%) Reversed

Autonomic Function
Sympathetic Nervous System tone Decreased (−40%) Reversed

Parasympathetic Nervous System tone Increased (80%) Unchanged

Neuroendocrine Function
Thyroid stimulating hormone Decreased (−18%) Unchanged
NIH-PA Author Manuscript

Triiodothyronine Decreased (−7%) Reversed

Thyroxine Decreased (−9%) Reversed

Gonadotropins Decreased Reversed

Circulating Leptin Decreased (proportional to fat mass) Reversed


NIH-PA Author Manuscript

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.
Rosenbaum and Leibel Page 15

Table 2

Studies of skeletal muscle in weight reduced subjects by ergometry, 31P-NMR spectroscopy, and analysis of
NIH-PA Author Manuscript

vastus lateralis muscle biopsy specimens 27, 97.

Ergometry 31P-NMR spectroscopy Biopsy

Efficiency NME ↑ 20% at low level exercise. 20% ↑ (Pi/PCr) at low level exercise.
18% ↓ ATP cost/muscle contraction.

Fuel Utilization 19% ↑ in % calories used derived 18% ↑ Pi (FFA/glucose oxidation potential). 12% ↓ PFK (glycolytic) activity,
from FFA oxidation during low No change kPCr (oxidative potential) 17% ↓ PFK/COX (glycolytic/FFA
level exercise oxidative activity)
No significant change in FFA
oxidative enzyme activities

Abbreviations: COX, cytochrome oxidase; kPCr, phosphocreatine recovery constant; FFA, free fatty acids; NMR, nuclear magnetic resonance;
PCr, phosphocreatine; PFK, phosphofructokinase; Pi, inorganic phosphate
NIH-PA Author Manuscript
NIH-PA Author Manuscript

Int J Obes (Lond). Author manuscript; available in PMC 2013 June 05.

You might also like